Sunteți pe pagina 1din 14

Journal of Microencapsulation, October 2008; 25(7): 499–512

Vesicular aceclofenac systems: A comparative study between


liposomes and niosomes

MAHA NASR1, SAMAR MANSOUR1, NAHED D. MORTADA1,


& A. A. ELSHAMY2
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

1
Department of Pharmaceutics and 2Department of Drug Technology, Faculty of Pharmacy,
Ain Shams University, Cairo, Egypt

(Received 15 July 2007; accepted 13 March 2008)

Abstract
Vesicular delivery systems have been reported to serve as local depot for sustained drug release. Aceclofenac multilamellar
liposomes and niosomes were prepared and a comparative study was done between them through evaluation of entrapment
efficiency, particle size, shape, differential scanning calorimetry and in vitro drug release. A stability study was carried out
by investigating the leakage of aceclofenac and the change in the vesicles particle size when stored at (2–8 C) for 3 months.
For personal use only.

The anti-inflammatory effect of aceclofenac vesicles was assessed by the rat paw oedema technique. Results showed that the
entrapment efficiency and the in vitro release of aceclofenac from the vesicles can be manipulated by varying the cholesterol
content, the type of surfactant as well as the type of charge. Niosomes showed better stability than liposomes. Both vesicular
systems showed significant sustained anti-inflammatory activity compared to the marketed product, with niosomes being
superior to liposomes as manifested by both oedema rate and inhibition rate percentages suggesting their effectiveness as topical
anti-inflammatory delivery systems.

Keywords: Aceclofenac, liposomes, niosomes, rat paw oedema, physical stability

Introduction valuable biocompatible drug delivery system (Pavelic


et al. 2001). Local anaesthetic agents, anti-
Aceclofenac is a phenylacetic acid derivative related to
inflammatory agents, anti-fungal agents, antibiotics,
diclofenac. It is a practically insoluble in water drug
vitamins and peptide or proteins are among the
with a molecular weight of 354.19, a pKa value of 4.7
substances whose liposomal application holds promise
and a log p-value of 1.23 (Alvarez-Larena et al. 1992; when applied topically for localized drug delivery
British Pharmacopoeia 1998). It is a potent analgesic, (Rolland 1993). However, despite that the majority of
anti-pyretic and anti-inflammatory agent used in the reports have concerned the use of phospholipid vesicles
management of moderate-to-severe pain and in or liposomes; they exhibit certain disadvantages, one of
rheumatic disorders such as rheumatoid arthritis and which relates to their chemical instability. The cost and
ankylosing spondylitis (Pasero et al. 1995; Martel- variable purity of natural phospholipids might lead
Pelletier et al. 1997). When taken orally, aceclofenac to other considerations. Therefore, alternatives to
induce the same side effects of other NSAIDs including phospholipids are the focus of interest from a technical
those related to GIT, liver and kidney function as well view point (Baillie et al. 1985).
as disturbance of platelets function (Insel 1992). Non-ionic surfactant vesicles (niosomes) have been
Vesicular delivery systems have proved superiority widely studied as an alternative to liposomes. They are
over conventional dosage forms, especially for the biodegradable, biocompatible, non-toxic and capable of
topical route of administration (Egbaria and Weiner encapsulating large quantities of material in a relatively
1991). Liposomal formulations have been widely small volume of vesicles. In fact they offer several
administered for topical treatment of diseases as a advantages over liposomes: higher chemical stability,

Correspondence: Maha Nasr, MSc, Faculty of Pharmacy, Department of Pharmaceutics, Ain Shams University, Cairo, Egypt. E-mail: maha2929@gmail.com

ISSN 0265–2048 print/ISSN 1464–5246 online ß 2008 Informa UK Ltd.


DOI: 10.1080/02652040802055411
500 M. Nasr et al.

Table I. Entrapment efficiency, particle size and T8h of multilamellar liposomes.

Liposomal formulations
(PC:CH) or (PC:CH:DP or SA) Entrapment efficiency* Distribution modal Polydispersity T8h2
(molar ratio) (%  SD) size (mm) (SPAN index)1 (%  SD)

PC:CH (7:4) 29.9  1 0.88 0.34 61.9  0.2


PC:CH (7:6) 31.8  0.6 0.70 0.43 53.5  1.4
PC:CH (7:7) 19.5  0.2 0.59 0.14 43.4  0.7
PC:CH:SA (7:4:1) 45.5  0.4 1.33 0.6 50.7  2.6
PC:CH:SA (7:6:1) 47.2  0.2 1.10 0.19 38.2  0.6
PC:CH:SA (7:7:1) 32.4  0.1 1.09 0.53 35.8  2
PC:CH:DP (7:4:1) 37.9  0.2 1.35 0.26 84.4  1.4
PC:CH:DP (7:6:1) 39.6  1 1.09 0.4 64.8  1.3
PC:CH:DP (7:7:1) 30.9  1.5 1.08 0.45 58.7  3

Notes: PC: phosphatidylcholine; CH: cholesterol; SA: stearylamine, DP: dicetylphosphate.


Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

*Each value is an average of three determinations.


1
Polydispersity/SPAN ¼ (particle diameter at 90% cumulative size – particle diameter at 10% cumulative size)/(particle diameter at 50%
cumulative size).
2
T8h: cumulative percentage released of aceclofenac after 8 h.

intrinsic skin penetration enhancing properties and Preparation of aceclofenac multilamellar liposomes
lower costs (Baillie et al. 1985; Uchegbu and
Aceclofenac multilamellar liposomal formulations were
Florence 1995; Uchegbu and Vyas 1998; Vora et al.
prepared using the thin film hydration technique
1998). Niosomes behave in vivo like liposomes, prolong-
(Gabriels and Plaizier-Vercammen 2003; Vyas et al.
ing the circulation of entrapped drug and altering its
2004). The composition of the formulations is shown
organ distribution and metabolic stability (Ruckmani
in Table I. The drug (50 mg) and the lipid components
For personal use only.

et al. 2000).
(200 mg), including the egg phosphatidylcholine (PC)
The aim of this work was to formulate aceclofenac and cholesterol (CH), either alone or mixed with charge
multilamellar liposomal and niosomal formulations inducing agents: stearylamine (SA) or dicetyl phosphate
using a thin film hydration method intended for topical (DP), in different molar ratios were accurately
administration on the skin. The prepared formulations weighed into a 250 ml long-neck quick fit round
were characterized and evaluated for their in vitro bottom flask and dissolved in chloroform:methanol
release performance. The anti-inflammatory activity of mixture (2:1, v/v). The organic solvent system was
selected formulations was evaluated and compared to slowly removed under reduced pressure, using a rotary
that of the marketed product. evaporator (Janke and Kunkel, model RVO5-ST, IKA
Laboratories, Staufen, Germany) at 40 C and 150 rpm
(Agarwal el al. 2001; Bhatia et al. 2004) such that a thin
Materials and methods film of dry lipids was formed on the inner surface of the
flask. The dry lipid film was slowly hydrated with 10 ml
Materials of phosphate buffered saline (PBS) pH 7.4 for 1 h.
Acelofenac was kindly provided by Bristol-Myers The resulting liposomal suspension was mechanically
Squibb Company (Cairo, Egypt). L-- Phosphatidyl shaken for 1 h using a thermostatically controlled
choline, type X-E: from dried egg yolk (PC), cholesterol mechanical shaker (Kottermann, Hanigsen, Germany)
(CH), stearylamine (SA), dicetyl phosphate (DP) and at 60 rpm and 40 C leading to the formation of
carrageenan were purchased from Sigma Chemical Co. multilamellar liposomes. The liposomal suspension
(St. Louis, USA). Sorbitan monopalmitate (Span 40) was left to mature overnight at 4 C, to ensure hydration
and sorbitan monostearate (Span 60) were purchased of the lipid.
from Fluka Chemical Co. (Buchs, Switzerland).
Methanol, chloroform, absolute ethyl alcohol, Preparation of aceclofenac multilamellar niosomes
potassium dihydrogen phosphate, disodium hydrogen Aceclofenac multilamellar niosomes were also prepared
phosphate and sodium chloride were purchased from using the thin film hydration method. The composition
Adwic, El-Nasr chemical Co. (Cairo, Egypt) according of the formulations is shown in Table II. Accurately
to the methods of Prolabo (Paris, France). Acetyl uranil weighed quantities of the drug (50 mg), the surfactant
(Uranyl acetate-2-hydrate) was purchased from Allied (either Span 40 or Span 60) and CH in different molar
Signal (Riedel-dahaen, Germany). Spectra/Por dialysis ratios were dissolved in chloroform/methanol mixture
membrane, 12.000–14.000 molecular weight cut-off, (2 : 1, v/v) in a round-bottom flask following the same
was purchased from Spectrum Laboratories Inc. procedure previously described for liposomes with the
(Rancho Dominguez, Canada). difference that the organic solvents were removed
Vesicularal aceclofenac systems 501

Table II. Entrapment efficiency, particle size and T8h of multilamellar niosomes.

Niosomal formulation Entrapment efficiency* Distribution modal Polydispersity T8h2


composition (molar ratio) (%  SD) size (mm) (SPAN index)1 (%  SD)

Span 40:CH (7:4) 27.7  0.6 0.70 0.38 58.2  3.2


Span 40:CH (7:6) 32.5  0.1 0.84 0.44 52.1  0.4
Span 40:CH (7:7) 14.2  0.2 0.84 0.43 46.1  1.4
Span 60:CH (7:4) 31.6  0.2 0.85 0.66 49.4  2.7
Span 60:CH (7:6) 45.1  0.5 0.89 0.14 43.3  2.2
Span 60:CH (7:7) 37.9  0.4 1.04 0.51 36.2  0.4

Notes: CH: cholesterol.


*Each value is an average of three determinations.
1
Polydispersity/SPAN ¼ (particle diameter at 90% cumulative size – particle diameter at 10% cumulative size)/(particle diameter at 50%
cumulative size).
2
T8h: cumulative percentage released of aceclofenac after 8 h.
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

under vacuum in the rotary evaporator at 60 C Transmission electron microscopy (Model
(Hao et al. 2002; Manconi et al. 2002). JEM–100S, Joel, Tokyo, Japan) was also used to
examine the vesicle ultrastructure (Maestrelli et al.
Determination of aceclofenac entrapment efficiency 2005). Negatively stained samples were prepared by
placing a drop of 1 ml of the vesicular suspension on a
Aceclofenac liposomes and niosomes were separated carbon coated grid (Ahn et al. 1995). The suspension
from free unentrapped drug by centrifugation at was left for 2 min, to allow its absorption in the carbon
13 000 rpm for 30 min. The pellets formed were film, and the excess liquid was drawn off with filter
washed twice each with 10 ml phosphate buffered paper. Subsequently, a drop of 2% (w/v) acetyl uranil
saline and recentrifuged again for 30 min (Morilla aqueous solution was placed on the grid. The excess
For personal use only.

et al. 2002). The prepared liposomal and niosomal was removed with distilled water and the samples were
formulations were decanted and kept in the refrigerator examined by TEM at 20 and 25 kV (Taylor et al. 1990;
(Skalko-Basnet et al. 2000). The concentration of the Nagarsenker et al. 1999; Manconi et al. 2003).
entrapped drug was determined by lysis of an aliquot of
100 ml of the liposomal or niosomal formulations with
Determination of vesicle size. The size of the prepared
absolute alcohol and sonication to obtain a clear
aceclofenac vesicles was determined by light
solution (Law et al. 1994; Fang et al. 2001a). The
scattering based on laser diffraction using the
concentration of aceclofenac in absolute alcohol was
Malvern Mastersizer (Malvern Instruments Ltd.,
determined spectrophotometrically at 276 nm using UV
Worcestershire, UK) (Joshi and Misra 2001). The
spectrophotometer (Shimadzu, model UV-1601 PC,
polydispersity, i.e. the width of the particle size
Kyoto, Japan). No interference from liposomal or
distribution was given by a SPAN index which was
niosomal components was found at this wavelength.
calculated by the following equation (Tabbakhian
Further dilution was made if necessary. The encapsula-
et al. 2006):
tion or entrapment efficiency was calculated through
the following relationship (Aggarwal and Kaur 2005):
Polydispersity ðSPANÞ ¼ ðD0:9  D0:1 Þ=D0:5 ð2Þ
Entrapment efficiency percentage
Entrapped drug where D0.9, D0.1 and D0.5 are the particle diameters
¼  100 ð1Þ
Total drug determined at the 90th, 10th and 50th percentile of
particles undersized, respectively.
Characterization of aceclofenac liposomes and niosomes
Photomicroscopic analysis and transmission electron Differential scanning calorimetry (DSC). DSC experi-
microscopy (TEM). Samples of the positively charged ments were performed with a differential scanning
aceclofenac liposomes composed of PC:CH:SA (7:4:1) calorimeter (Schimadzu, model TA-50 WSI, Kyoto,
molar ratio and aceclofenac niosomes composed of Japan) calibrated with indium. Samples of aceclofenac,
Span 60:CH (7:4) molar ratio were used. plain and drug loaded multilamellar liposomes
For microscopic investigation, the vesicles were composed of PC:CH:SA (7:6:1) molar ratio as well as
examined under a binocular optical microscope multilamellar niosomes of Span 60:CH (7:6) molar ratio
(Carl Zeiss, Berlin, Germany) and photographed at a were submitted to DSC analysis. The analyses were
magnification of 400 (Gabriels and Plaizier- carried out on 40 ml or 1 mg samples sealed in
Vercammen 2003) by means of a fitted camera standard aluminum pans. Thermograms were
(Panasonic, Japan) for morphological evaluation. obtained at a scanning rate of 10 C min1 using dry
502 M. Nasr et al.

nitrogen flow (25 ml min1). Isotonic phosphate buffer respectively, Group V received aceclofenac liposomes
(pH 7.4) was employed as a reference (Ganesan et al. composed of PC:CH:SA (7:6:1) molar ratio, respec-
1984; Fresta et al. 1998). tively, Group VI received aceclofenac niosomes com-
posed of Span 60:CH (7:4) molar ratio, respectively,
In vitro release of aceclofenac from liposomes and Group VII received aceclofenac niosomes composed of
niosomes. The release of aceclofenac from liposomal Span 60:CH (7:6) molar ratio, respectively, and Group
and niosomal formulations was determined using the VIII received the marketed Bristaflam creamÕ . The
membrane diffusion technique (Devaraj et al. 2002; volume of paw oedema (ml) was measured in each
Glavas-Dodov et al. 2002). An accurately measured animal using a plethysmometer (UGO-Basile, 7140,
amount of aceclofenac vesicular formulations, equiva- Comerio, Italy) to a precision of two decimal places.
lent to 2 mg aceclofenac, was suspended in 1 ml PBS The rats were marked on the left hind paw just beyond
pH 7.4. The suspension was transferred to a glass the tibiotarsal junction, so that every time the paw was
cylinder having a length of 7 cm and diameter of 2.5 cm dipped in the electrolyte fluid column up to a fixed
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

previously covered at its lower end with a soaked mark to ensure constant paw volume.
cellulose membrane (Spectra/Por dialysis membrane The tested formulations were applied to the left hind
12–14 000 Mw cut-off). The cylinder was placed in the paws of rats using an amount from each of them
dissolution flask of a USP dissolution tester (Pharma equivalent to 1 mg of aceclofenac. The area of applica-
Test, Hainburg, Germany) containing 100 ml PBS pH tion was occluded with a parafilm (Nagarsenker and
7.4. The cylinder was allowed to rotate at a constant Joshi 1997) and was left in place for 2 h. The parafilm
speed (50 rpm). The phosphate buffered saline was was then removed and the residual formula on the
maintained at a temperature of 32 C. At pre-deter- surface was wiped off (Shahiwala and Misra 2002).
mined time intervals (0.25, 0.5, 1, 2, 3, 4, 5, 6, 7 and 8 h), After 2 h of topical application, initial paw volume of
samples were withdrawn and assayed spectrophotome- the rats was measured by dipping the rat paw into the
trically at 274 nm for the drug content. Each withdrawn electrolyte column. A volume of 0.1 ml of 1% (w/v)
sample was replaced by an equal volume of PBS carrageenan solution in saline was then injected in the
For personal use only.

(Ruckmani et al. 2000). The results were the mean of sub-plantar region of left hind paw of the rats and the
three runs. The obtained data were kinetically treated to increase in volume due to fluid displacement was noted
determine the order of release. from a digital display (Penzes et al. 2005). Measurement
of paw volume was done after 1, 2, 3, 4, 5, 6, 7 and 8 h.
Physical stability. The selected liposomal and nioso- The oedema and inhibition rate percentage of each
mal batches were sealed in 20 ml glass vials and stored group was calculated as follows:
as pellets (2–8 C) (Patel and Misra 1999). Samples from
each batch were withdrawn at specified time intervals to Vt  V0
Oedema rate ðE%Þ ¼  100 ð3Þ
determine the residual amount of the drug in the V0
vesicles as described previously (Shahiwala and Misra
2002; Sinico et al. 2005). The leaked out drug during the Ec  Et
Inhibition rate ðI%Þ ¼  100 ð4Þ
storage period was separated by dilution of an aliquot Ec
of each formulation with phosphate buffered saline and
where V0 is the mean paw volume before carrageenan
centrifugation at 13 000 rpm for 30 min. At the end of
injection (ml), Vt is the mean paw volume after
the storage period, particle size was also determined at
carrageenan injection (ml), Ec is the oedema rate of
the end of the storage period; results of which can be
control group and Et is the oedema rate of the treated
taken as an index of stability (Yan-yu et al. 2006).
group (Mei et al. 2003, 2005).
Data were expressed as mean  SD and statistically
In vivo study of liposomes and niosomes. This work will
assessed by one way analysis of variance (ANOVA).
include the estimation of the anti-inflammatory activity
Values for oedema rate percentage for liposomal
of the selected aceclofenac liposomes and niosomes
formulations, niosomal formulations and the marketed
compared to plain vesicles as well as to the marketed
product were compared to the saline control and the
product using the rat paw oedema test. The protocol of
differences were determined statistically using
the present work was approved by Experiments
Dunnett’s t-test.
and Advanced Pharmaceutical Research Unit
(EAPRU), Faculty of Pharmacy, Ain Shams
University, Cairo, Egypt.
Adult male albino rats, weighing 130–150 g, were Results and discussion
divided into eight groups of five animals each, namely:
Entrapment efficiency for liposomal formulations
Group I received topical saline application, Group II
received plain liposomes, Group III received plain Effect of cholesterol content. Egg phospholipids were
niosomes, Group IV received aceclofenac liposomes chosen rather than soya phospholipids for the
composed of PC:CH:SA (7:4:1) molar ratio, preparation of liposomes because previous results
Vesicularal aceclofenac systems 503

have shown that they exhibited significantly higher Entrapment efficiency for niosomal formulations
hydration effects than soya phospholipids; therefore,
Cholesterol content. Data in Table II shows that the
they are recommended for the preparation of topical
entrapment efficiency percentages of aceclofenac
formulations (Betz et al. 2005).
in niosomal formulations were 27.7%, 32.5% and
Data in Table I reveals that increasing the CH
14.2% for multilamellar niosomes composed of 7:4,
content in liposomal formulations significantly
7:6 and 7:7 Span 40:CH molar ratios, respectively.
increased the entrapment efficiency of aceclofenac up
On the other hand, the percentages of entrapment
to an optimum PC:CH ratio (7:6). The percentage
efficiency of aceclofenac were 31.6%, 45.1%, 37.9% for
entrapment efficiency significantly increased from
multilamellar niosomes composed of 7:4, 7:6 and 7:7
29.9% to 31.8% for neutral liposomes composed of
Span 60:CH molar ratios, respectively. As observed
7:4 and 7:6 PC:CH molar ratios, respectively, but
with liposomal formulations, the incorporation of
it significantly decreased to 19.5% with liposomal
CH into niosomes significantly increased the drug
formulations of 7:7 PC:CH molar ratio (p 5 0.05).
entrapment efficiency up to an optimum Span:CH
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

The increase in entrapment efficiency is explained on


ratio (7:6) (p 5 0.001). Cholesterol alters the fluidity of
the basis of recognized effects for cholesterol: increasing
chains in bilayers and, when present in sufficient
the microviscosity and rigidity of the lipidic bilayer,
concentration, abolishes the gel to liquid phase
resulting in a higher stability (Moribe et al. 1999),
transition of surfactant bilayers (Uchegbu and
reduced permeability (Du Plessis et al. 1996) of the
Florence 1995; Devaraj et al. 2002, Hao et al. 2002).
liposomal membrane and hence greater drug retention It also increases the microviscosity of niosomal
(Joshi and Misra 2001). However, further increase in membrane conferring more rigidity (Manosroi et al.
CH content beyond a certain level (7:6 PC:CH molar 2003). Further increase of CH content to reach 7:7
ratio) starts disrupting the regular bilayered structure Span:CH molar ratio significantly reduced the entrap-
leading to lowering of drug entrapment levels (Patel and ment efficiency. This could be interpreted similarly as in
Misra 1999). the case of aceclofenac liposomes based on the effect of
cholesterol on the rigidity of the regular bilayered
For personal use only.

Charge inducing agents. Stearylamine and dicetyl structure leading to loss of drug entrapment levels
phosphate were used as positive and negative charge (Agarwal el al. 2001; Guinedi et al. 2005).
inducers, respectively. Data in Table I reveals that the
charged liposomes exhibited an increased entrapment The type of surfactant. An increasing number of
of aceclofenac compared to neutral ones (Sharma et al. non-ionic surfactants have been found to formulate
1994). The percentages encapsulation efficiencies niosomes among which sorbitan esters are much more
significantly increased from 29.9% for neutral common. It was reported that the length of alkyl chain
liposomes composed of PC:CH (7:4) molar ratio to is a crucial factor of permeability and that long chain
45.5% and 37.9% for their positively and negatively surfactants produce high drug entrapment into vesicles
charged counterparts, respectively, from 31.8% for (Hao et al. 2002). Therefore, in this study Span 40 and
neutral liposomes of PC:CH (7:6) molar ratio to 47.2% Span 60 were chosen as the surfactants of choice for the
and 39.6% for their positively and negatively charged formation of the non-ionic vesicles.
counterparts, respectively, and from 19.5% for neutral By inspection of data in Table II it can be concluded
liposomes composed of PC:CH (7:7) molar ratio to that the entrapment efficiency percentage for niosomes
32.4% and 30.9% for their positively and negatively prepared using Span 60 were significantly higher
charged counterparts, respectively (p 5 0.05). (p 5 0.001) than those prepared using Span 40 with
The incorporation of charged lipids into bilayers the same molar ratios of Span:CH. This can be
increases the volume of the aqueous compartments by explained by the fact that Span 60 has higher phase
separating adjacent bilayers due to charge repulsion, transition temperature (Yoshioka et al. 1994). It was
resulting in an increase in the interlamellar resistance reported that the higher the transition temperature of
between successive bilayers and an increase in trapped the surfactant the higher the encapsulation efficiency
volume, hence increase in encapsulation (Kulkarni (Uchegbu and Vyas 1998). Long chain surfactant
et al. 1995; DuPlessis et al. 1996). Moreover, data in produces high entrapment (Hao et al. 2002) and it has
Table I reveals that positively charged liposomes been demonstrated that entrapment efficiency may be
exhibited the highest entrapment efficiency followed correlated to the hydrophobicity of the alkyl chain of
by negatively charged ones then neutral ones, using the the sorbitan esters (Manconi et al. 2002). Hence, Span
same PC:CH molar ratio. Aceclofenac is a weak acid 60 having longer saturated alkyl chain (C18) compared
and an electrostatic attraction would occur between to Span 40 (C16) produced niosomes with higher
drug anion and the positively charged SA, this entrapment efficiency. In addition, the length of the
attraction would account for the highest entrapment alkyl chain influences the HLB value of the surfactant
efficiency obtained with aceclofenac positively charged mixture which in turn directly affects the drug
liposomal formulations (El-Gazayerly and Hikal 1997). entrapment efficiency. The HLB values for Span 40
504 M. Nasr et al.

(a) (b)

Figure 1. Negative stain electron micrograph of (a) aceclofenac liposomes composed of PC:CH:SA (7:4:1) molar ratio and
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

(b) aceclofenac niosomes composed of Span 60:CH (7:4) molar ratio.

and Span 60 are 6.7 and 5, respectively, as provided by increased the spacing between adjacent bilayers by
the manufacturer, the lower the HLB of the surfactant repulsion (Taylor et al. 1990; Vyas et al. 1995), thus
the higher will be the drug entrapment efficiency increasing the size of the internal aqueous compartment
(Guinedi et al. 2005). resulting in the formation of liposomes larger in size
By comparing the entrapment efficiency percentages compared to the neutral ones. Furthermore, the
of the prepared aceclofenac liposomal and niosomal positively charged lipid electrostatically attracts the
formulations, it can be concluded that aceclofenac drug anion, which may be expected to push phospho-
niosomes prepared from Span 60:CH (7:6) molar ratio lipids headgroups apart, thereby increasing the particle
showed higher entrapment efficiency than liposomes diameter (Hathout et al. 2007). This increase in particle
For personal use only.

composed of PC:CH of the same molar ratio. This size would account for the higher entrapment efficiency
could be ascribed to the solubilizing effect of the of the charged liposomes compared to the neutral ones
surfactant used in the preparation of niosomes on (Vyas et al. 1995).
aceclofenac. Data in Table II reveals that niosomes prepared using
Span 60 were slightly larger in size than those prepared
Characterization of aceclofenac liposomes and using Span 40 (Manconi et al. 2002; Guinedi et al.
niosomes 2005). Span 60 has a longer saturated alkyl chain
Photomicroscopic analysis and transmisson electron compared to Span 40, which could increase the size of
microscopy. The photomicrographs of aceclofenac the vesicle (Manosroi et al. 2003). Larger vesicles are
multilamellar liposomes composed of PC:CH:SA formed when the hydrophilic portion of the molecule is
(7:4:1) molar ratio and multilamellar niosomes of decreased relative to the hydrophobic portion, so this
Span 60:CH (7:4) molar ratio revealed the spherical would account for the higher entrapment efficiencies
shape and the multilayered structure of the prepared obtained with Span 60 multilamellar niosomes
liposomes and niosomes that exist in disperse and in (Uchegbu and Duncan 1997), it may also be
aggregate collections (results not shown). Spherical attributed to the fact that the increase in alkyl chain
niosomes or liposomes display an elastic memory which length as the series is ascended from the C12 to the C18
has been argued to be of paramount importance for the ester would result in an increase in the value of the
success of the carrier-mediated material transport critical packing parameter and hence the minimal
across the skin (Nasseri 2005). permissible vesicle size would increase (Uchegbu and
The electron micrographs of the previous samples are Florence 1995).
shown in Figures 1(a) and (b), respectively. They show Regarding polydispersity index, a value of 0 indicates
the outline and the core of the well identified multi- an entirely monodisperse population and a value of 1
lamellar vesicles displaying the retention of sealed indicates a completely polydisperse population
vesicular structure (Morilla et al. 2002). (Zeisig et al. 1996). The prepared vesicular formulations
were mostly heterogenous, which is usually obtained
Vesicle size analysis. Investigation of Table I reveals with vesicles prepared using the thin film hydration
that charged liposomes exhibited mean particle method not followed by sonication or extrusion of the
diameter higher than that of the neutral ones. The vesicles (Pereira-Lachataignerais et al. 2006).
frequency distribution curves of particle size data were The particle size of the prepared vesicles is well suited
unimodal in shape and demonstrated log-normal for topical delivery through the skin as particles smaller
distribution of particle size (Manosroi et al. 2002). than 3 mm were found to be distributed into the stratum
The inclusion of a charge inducer in liposomes corneum and hair follicles (Friedman et al. 1995).
Vesicularal aceclofenac systems 505
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13
For personal use only.

Figure 2. DSC thermograms of aceclofenac, plain and drug loaded positively charged liposomes composed of PC:CH:SA
(7:6:1) molar ratio.

Differential scanning calorimetry (DSC). DSC Cholesterol incorporated into the liposomal membrane
thermograms of aceclofenac, plain and drug-loaded had its hydroxyl group oriented towards the aqueous
multilamellar liposomes composed of 7:6:1 PC:CH:SA layer, with the aliphatic chain aligned parallel to the
molar ratio as well as multilamellar niosomes composed acyl chains in the centre of lipid bilayers. The presence
of Span 60:CH (7:6) molar ratio are illustrated in of rigid steroid nuclei alongside the carbons of the
Figures 2 and 3, respectively. These formulations phosphatidylcholine chain may reduce the freedom of
represented the highest entrapment efficiency among movement. This may result in an enhanced stability of
the prepared liposomes and niosomes, respectively. the liposomal membrane (Manosroi et al. 2002).
A DSC thermogram of aceclofenac showed an A DSC thermogram of plain niosomes composed of
endothermic peak at 150.4 C, which is the reported 7:6 Span 60:CH molar ratio showed an endothermic
melting point of the drug in the literature. (Budavari peak at 105.5 C. A DSC thermogram of aceclofenac-
et al. 1996). A DSC thermogram of plain liposomes loaded niosomes of the same Span 60:CH molar ratio
containing PC:CH:SA in the molar ratio of (7:6:1) showed disappearance of the melting endotherm of
showed a peak endotherm at 102 C. A DSC aceclofenac and broadening of the endothermic peak
thermogram of aceclofenac-loaded liposomes at 103.4 C.
composed of PC:CH:SA (7:6:1) molar ratio interest- To recapitulate, the absence of the melting
ingly showed disappearance of the melting endotherm endotherm of aceclofenac and shifting and/or broad-
of aceclofenac and the shift of the endotherm from ening of the endotherms of the lipid bilayer components
102 C to 92.3 C. This may suggest that aceclofenac of liposomes or surfactant bilayers of niosomes
can fit into the phospholipids bilayer producing suggests significant interaction of aceclofenac with
significant perturbation in the packing characteristics bilayer components and can account for the enhanced
of the phospholipid membrane and accordingly was entrapment of aceclofenac into these formulations
able to reduce the peak of the main transition (Fresta (Nagarsenker and Joshi 1997; Hathout et al. 2007).
and Puglisi 1997; Maghraby et al. 2005).
Plain and drug loaded liposomal formulations
In vitro drug release studies
showed broad transitions which are characteristic for
lipid mixtures containing cholesterol, signifying good In vitro release of aceclofenac from liposomes. The
interaction of all components forming the bilayers of percentages of drug released after 8 h (T8h) were 61.9%,
liposomes (Blume et al. 1993; Nagarsenker et al. 1999). 53.5% and 43.4% for multilamellar liposomal
506 M. Nasr et al.
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13
For personal use only.

Figure 3. DSC thermograms of aceclofenac, plain and drug loaded niosomes composed of Span 60:CH (7:6) molar ratio.

formulations composed of 7:4, 7:6 and 7:7 PC:CH (7:4:1, 7:6:1 and 7:7:1) molar ratios, respectively, were:
molar ratios, as shown in Table I. Therefore, on the 84.4%, 64.8% and 58.7%, with significant differences
basis of the molar ratio of the lipid content, T8h for at p 5 0.05. Therefore, the order of release of
the neutral liposomal preparations can be arranged in aceclofenac from multilamellar liposomes may be
the following decreasing order: 7:4 4 7:6 4 7:7 with arranged in the following order: negatively charged
significant differences at p 5 0.001. The results could be liposomes 4 neutral liposomes 4 positively charged
due to the fact that the presence of CH in the bilayers at liposomes. This order may be due to the electrostatic
a temperature above the transition temperature (Tc) attraction forces that may exist between the anionic
modulates membrane fluidity by restricting the drug and positively charged liposomes and vice versa
movement of the relatively mobile hydrocarbon for negatively charged ones.
chains, hence reducing bilayer permeability
(Nagarsenker and Londhe 2003), and condenses the In vitro release of aceclofenac from niosomes. From
packing of the phospholipids in the bilayers (Arica et al. data in Table II it is obvious that the increase of CH
1995), thus decreasing the efflux of the encapsulated molar ratio from 7:4 to 7:6 and 7:7 significantly reduced
drug (Zhang and Zhu 1999; Fatouros et al. 2001), the efflux of aceclofenac, which is in accordance with
resulting in prolonged drug retention (Taylor cholesterol membrane stabilizing ability and space
et al. 1990). filling action (Uchegbu and Florence 1995; Namedo
Data in Table I also shows the effect of charge and Jain 1999). Cholesterol is known to increase the
inducers on T8h of aceclofenac from multilamellar rigidity of niosomes (Vyas and Venkatesan 1999) and
liposomal formulations with the lipid content PC:CH abolish the gel-to-liquid phase transition of niosomal
7:4, 7:6 and 7:7 molar ratios. Positively charged systems resulting in niosomes that are less leaky
liposomes showed the lowest T8h, where values were (Rogerson et al. 1987), thus decreasing the drug release
50.7%, 38.2% and 35.8% for liposomal formulations from niosomes (Alsarra et al. 2005). Lipophilic drugs
composed of 7:4:1, 7:6:1 and 7:7:1 PC:CH:SA molar are known to be intercalated within the bilayer
ratios, respectively. In contrast, negatively charged structure of niosomes (Fang et al. 2001b), thus the
liposomes exhibited the highest rates and extent of release of aceclofenac is sustained from niosomes.
drug release. The percentages of drug released after 8 h T8h of aceclofenac was 58.2%, 52.1% and 46.1% for
for liposomal formulations composed of PC:CH:DP 7:4, 7:6 and 7:7 Span 40:CH molar ratios, respectively,
Vesicularal aceclofenac systems 507

Table III. Determination of the order of release of aceclofenac from different liposomal and niosomal formulations using the coefficient
of determination (r2).

r2

Liposomal or niosomal formulation composition (molar ratio) Zero order First order Diffusion model

PC:CH (7:4) 0.913 0.962 0.986


PC:CH:SA (7:4:1) 0.956 0.979 0.991
PC:CH:DP (7:4:1) 0.928 0.986 0.989
PC:CH (7:6) 0.972 0.986 0.988
PC:CH:SA (7:6:1) 0.953 0.972 0.988
PC:CH:DP (7:6:1) 0.941 0.981 0.988
PC:CH (7:7) 0.969 0.987 0.991
PC:CH:SA (7:7:1) 0.940 0.960 0.988
PC:CH:DP (7:7:1) 0.885 0.930 0.964
Span 40:CH (7:4) 0.901 0.953 0.970
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

Span 40:CH (7:6) 0.946 0.973 0.992


Span 40:CH (7:7) 0.925 0.956 0.984
Span 60:CH (7:4) 0.894 0.934 0.977
Span 60:CH (7:6) 0.899 0.932 0.980
Span 60:CH (7:7) 0.947 0.964 0.991

Note: PC: phosphatidylcholine; CH: cholesterol; SA: stearylamine, DP: dicetylphosphate.

while the T8h for 7:4, 7:6 and 7:7 Span 60:CH molar
ratios were 49.4%, 43.3% and 36.2%, respectively. On
the basis of Span:CH ratio, T8h can be arranged in the
following decreasing order: 7:4 4 7:6 4 7:7.
Niosomal formulations prepared using Span 60
For personal use only.

exhibited a significantly slower rate of drug release


compared to Span 40. This can be explained by the fact
that niosomes exhibit an alkyl chain length-dependent
release. The higher the chain length, the lower the
release rate (Devaraj et al. 2002).
Linear regression analysis of the release data reveals
that the drug is released from liposomes and niosomes
by a diffusion controlled mechanism, as shown in Figure 4. Effect of storage on the percentage aceclofenac
Table III. These results are in agreement with many retained in the prepared liposomal and niosomal vesicles.
research workers who found that the drugs were
released from vesicles by the same mechanism Effect of storage on drug leakage from
(Arica et al. 1995; Glavas-Dodov et al. 2002; Hathout vesicular systems
et al. 2007).
Drug leakage from the vesicles was evaluated after 15,
30, 45, 60 and 90 days and the results are illustrated in
Physical stability studies Figure 4 in terms of percentage of aceclofenac retained
Positively charged aceclofenac liposomal formulations in the vesicular formulations (Patel and Misra 1999).
composed of PC:CH:SA of 7:4:1 and 7:6:1 molar ratios The percentages of aceclofenac retained in the liposo-
and niosomal formulations composed of Span 60:CH of mal formulations after 90 days were 51% and 63% for
7:4 and 7:6 molar ratios were considered in this study liposomal formulations composed of PC:CH:SA (7:4:1)
as they represented formulations of the highest entrap- and PC:CH:SA (7:6:1) molar ratios, respectively. The
ment efficiency. Physical stability studies on selected leakage of the drug from liposomes may be attributed
aceclofenac liposomal and niosomal formulations to many reasons such as: the fluidity and mobility of the
stored at refrigeration temperature (2–8 C) for a phospholipid bilayer of the liposomes, the tightness of
period of 3 months were conducted by monitoring (a) interaction of the entrapped materials to the phospho-
the leakage of encapsulated aceclofenac from liposomes lipid bilayers and the size of entrapped molecules
and niosomes and (b) the change in particle size related to their penetrability to the bilayer membrane
distribution of the vesicles after storage. In previous (Law et al. 1994).
studies pellets of the vesicles were selected for this study Moreover, The percentage of aceclofenac retained in
as they showed low leakage rates when stored in the the case of liposomes composed of PC:CH:SA (7:6:1)
refrigerator under iso-osmotic conditions (Ozer and was significantly more than those composed of
Talsama 1989). PC:CH:SA (7:4:1) at p 5 0.001; this may be attributed
508 M. Nasr et al.

Table IV. Effect of 3 months storage on particle size distribution of selected liposomal and niosomal formulations.

Distribution modal size (mm)

Vesicle type Vesicle composition (molar ratio) Fresh vesicles Stored vesicles

Liposomes PC:CH:SA (7:4:1) 1.33 (0.66, 1.6)


PC:CH:SA (7:6:1) 1.11 (0.83, 1.29)
Niosomes Span 60:CH (7:4) 0.85 0.86
Span 60:CH (7:6) 0.89 1.08

to the fact that the presence of cholesterol in the bilayer The degradation of non-ionic surfactant is limited so
of the egg phosphatidylcholine liposomes dramatically the replacement of lecithin by non-ionic surfactant has
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

reduced the leakage of encapsulated material been considered to overcome this problem. Also the
(Crommelin et al. 1994) by decreasing the rotational entrapment of hydrophobic drugs in niosomes is known
freedom of the phospholipid hydrocarbon chains to increase their stability (Sihorkar and Vyas 2000).
(Betageri 1993). This confers structural stability and Previous work has also demonstrated that Span 60
gives a more ordered structure bilayer (Patel and niosomal formulations are generally very stable
Misra 1999). (Gianasi et al. 1997) and that drug leaching from
On the other hand, the percentages of aceclofenac niosomal vesicles made from Span is low due to its high
retained after a period of 3 months in multilamellar phase transition temperature and its low permeability
niosomal formulations composed of Span 60:CH of 7:4 (Vora et al. 1998).
and 7:6 molar ratio were 63.4% and 72.4%, The previous results confirm that niosomes exhibit
respectively. Aceclofenac was significantly more superior stability as compared to liposomes (Vora et al.
retained in Span 60 niosomes of 7:6 than 7:4 molar 1998) manifested by better entrapment efficiency and
For personal use only.

ratio (p 5 0.05), due to the membrane stabilizing ability slight changes in particle size.
of cholesterol, thus reducing the leakage of drug. The
results indicate that the percentages of aceclofenac In vivo anti-inflammatory study
retained in niosomal formulations were higher than
liposomal counterparts. The liposomal formulations composed of PC:CH:SA of
7:4:1 and 7:6:1 molar ratio and niosomal formulations
composed of Span 60:CH of 7:4 and 7:6 molar ratio
Effect of storage on particle size distribution of
were selected as they represented the formulations
vesicular formulations
with the highest drug entrapment. Positively charged
The effect of storage on particle size of liposomal and liposomes were chosen for the in vivo study for its well
niosomal formulations is demonstrated in Table IV. known adhesion to the negatively charged skin, as
The results showed (a) slight increase in the vesicle reported by Manosroi et al. (2004).
diameters of the stored liposomal formulations and From the results demonstrated in Table V, it can
(b) slight change in the frequency distribution curves be concluded that there was overall non-significant
which was found to be bimodal. This may be due to differences in the oedema rate percentage results
slight fusion and aggregation of the liposomes after between group I (the normal saline control) and
storage (Padamwar and Pokharkar 2006). Charge on group II (the plain liposomes) or group III (the plain
lipids is an important parameter influencing liposomal niosomes) at p 5 0.05, suggesting that the plain
behaviour. In this way, high surface potential might liposomes or niosomes do not possess in vivo anti-
increase liposome physical stability by reducing the rate inflammatory effect (Naresh et al. 1994). Differences in
of aggregation and fusion due to electrostatic repulsion oedema rate percentages between plain liposomes or
(Du Plessis et al. 1996; Padamwar and Pokharkar niosomes and the saline control were determined using
2006). Niosomal formulations also showed a slight an unpaired t-test.
change in the size and distribution curves on storage Oedema rate of the left foot pad reached its peak at
(Gianasi et al. 1997) but this was found to be minimal 4 h. The oedema rate and inhibition rate percentages
compared to liposomes indicating a much more obtained with different tested preparations are shown in
stable system. Table V. All the tested formulations showed better anti-
Comparing the results of the physical stability study inflammatory activity than saline control.
of liposomes to that of niosomes, it could be concluded Aceclofenac liposomal and niosomal formulations
that niosomes gave better stability. This is possibly showed significant (p 5 0.05) percentage inhibition of
because the problems of phospholipid hydrolysis and oedema compared to control at all time intervals,
fatty acid peroxidation in liposomes were not observed suggesting the sustained anti-inflammatory activity of
for non-ionic surfactants (Fang et al. 2001a). aceclofenac vesicles throughout the whole time of the
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13
For personal use only.

Table V. Oedema rate percentages of plain and aceclofenac-loaded liposomes and niosomes.

Odema rate (%)  SD [Inhibition rate (%)]

Tested formulation 1h 2h 3h 4h 5h 6h 7h 8h

Control saline (Group I) 50.1  8.5 53.3  3.2 63.5  5.7 66.1  3.8 59.3  6.4 52.3  1.9 48.7  3.8 46.4  3.6
Plain liposomes (Group II) 43.2  1.7 51.1  2 60.3  2.7 63.5  0.6 61.6  4.2 59.5  5 48.8  6 40.2  4.9
Plain niosomes (Group III) 50  0.5 54.5  3.4 64.7  1.8 69.7  1.9 56.8  5.4 55.3  1.2 51.1  6.7 39.2  3.5
PC:CH:SA (7:4:1) liposomes (Group IV) 35.7  4.7 38.3  4.5 45  3.6 49.6  0.6 45.8  3.5 44.1  3.9 35.2  3.1 33.1  3.9
[28.7] [28.1] [29.2] [25] [22.8] [15.7] [27.8] [28.6]
PC:CH:SA (7:6:1) liposomes (Group V) 36.1  5.1 37.4  0.5 48.7  5.4 56.2  2.6 52.1  2.6 49  1.6 40.2  0.9 38.7  1.5
[28] [29.9] [23.3] [15] [12.3] [6.3] [17.4] [16.7]
Span 60:CH (7:4) niosomes (Group VI) 34.1  4.7 34.2  1.6 49.7  1.4 50.8  3.4 38.7  3.3 37.4  1.8 31.3  0.8 27.2  3.1
[32] [35.9] [21.7] [23.1] [34.7] [28.5] [35.7] [41.4]
Span 60:CH (7:6) niosomes (Group VII) 34.9  1.9 35  2.9 46.5  2.8 49.1  0.8 44.7  4.8 44.1  2 41.7  2.3 33.1  0.4
[30.5] [34.3] [26.8] [25.7] [24.7] [15.7] [14.3] [28.6]
Marketed product (Group VIII) 26.8  1.9 31.5  2.9 38  1.4 51.1  1.1 40.2  2.3 40.4  1.6 41.3  2.9 42  5.2
[46.5] [40.9] [40.1] [22.7] [32.2] [22.7] [15.2] [9.6]

Statistically significant from saline control Dunnett’s t-test; p 5 0.01, p 5 0.05.
PC: phosphatidylcholine, CH: cholesterol, SA: stearylamine.
Vesicularal aceclofenac systems
509
510 M. Nasr et al.

study (8 h). Liposomes and niosomes prepared using Ahn BN, Kim SK, Shim CK. 1995. Preparation and evaluation
of proliposomes containing propranolol hydrochloride.
PC:CH and Span:CH at a molar ratio of 7:4 showed
J Microencapsulation 12:363–375.
better anti-inflammatory activity than those prepared Alsarra IA, Bosela AA, Ahmed SM, Mahrous GM. 2005.
with the 7:6 molar ratio. This may be attributed to the Proniosomes as a drug carrier for transdermal delivery of
fact that, by increasing the CH content, a condensing ketorolac. Eur J Pharm Biopharm 59:485–490.
effect on the vesicle membranes was exerted, making Alvarez-Larena A, Piniella JF, Carrasco E, Ginebreda A, Julia S,
Germain G. 1992. Crystal structure and spectroscopic study of
them more rigid with a decreased rate of drug
2-[(2,6-dichlorophenyl) amino] phenylacetoxyacetic acid
permeation across the skin. (Aceclofenac). J Crystallographic Spectroscopic Res 22:323–328.
By comparing vesicular formulations to the marketed Arica B, Ozer AY, Ercan MT, Hincal AA. 1995. Characterization,
product, it was obvious that liposomal and niosomal in vitro and in vivo studies on primaquine diphosphate liposomes.
tested formulations produced significantly higher anti- J Microencapsulation 12:469–485.
Baillie AJ, Florence AT, Hume LR, Muirhead GT, Rogerson A.
inflammatory activity (lower oedema rate percentage)
1985. The preparation and properties of niosomes- non-ionic
after 6 h and 3 h, respectively. The effect was sustained surfactant vesicles. J Pharm Pharmacol 37:863–868.
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

until the end of the experiment (8 h), contrary to the Betageri GV. 1993. Liposomal encapsulation and stability of
marketed product. It was reported that the vesicle dideoxyinosine triphosphate. Drug Dev Ind Pharm 19:531–539.
components together with the drug are deposited into Betz G, Aeppli A, Menshutina N, Leuenberger H. 2005. In vivo
comparison of various liposome formulations for cosmetic
the skin, thus creating a microreservoir from which the application. Int J Pharm 296:44–54.
drug is slowly released (El Ridy and Khalil 1999; Bhatia A, Kumar R, Katare OP. 2004. Tamoxifen in topical
Shahiwala and Misra 2002). liposomes: Development, characterization and in-vitro evaluation.
It was obvious that niosomes showed superior anti- J Pharm Pharm Sci 7:252–259.
inflammatory activity than liposomes starting from the Blume A, Janesan M, Ghyczy M, Gareiss J. 1993. Interaction of
phospholipid liposomes with lipid model mixtures for stratum
fifth hour. This is in accordance with other studies corneum lipids. Int J Pharm 99:219–228.
which revealed that surfactant treated formulations British Pharmacopoeia, Vol. I. 1998. London: Her Majesty’s
were superior to phospholipid treated formulations in Stationary Office. p 33.
facilitating the permeation of drugs, especially those Budavari S, O’Neil MJ, Smith A, Heckelman PE, Kinneary JF,
For personal use only.

prepared using Span 60 (Fang et al. 2001b). Niosomes editors. 1996. The Merck Index, an encyclopedia of chemicals,
drugs and biological. 12th ed. Whitehouse station, NJ: Merck and
loaded with drugs for dermal application are aimed to Co., Inc. p 5.
preferentially show interactions with the inflamed Crommelin DJA, Grit M, Talsama H, Zuidam NJ. 1994. Liposomes
(epidermal) tissue without exerting an immediate or as carriers for drugs and antigens: Approaches to preserve their
strong systemic action (Junginger et al. 1991; Shahiwala long term stability. Drug Dev Ind Pharm 20:547–556.
Devaraj GN, Parakh SR, Devraj R, Apte SS, Rao BR, Rambhau D.
and Misra 2002).
2002. Release studies on niosomes containing fatty alcohols as
bilayer stabilizers instead of cholesterol. J Coll Interf Sci
251:360–365.
Du Plessis J, Ramachandran C, Weiner N, Muller DG. 1996.
Conclusion The influence of lipid composition and lamellarity of liposomes on
the physical stability of liposomes upon storage. Int J Pharm
The liposomal and niosomal formulations proved to 127:273–278.
be promising topical systems for the delivery of Egbaria K, Weiner N. 1991. Topical application of liposomal
aceclofenac as they provided a consistent and preparations. Cosmetics Toiletries 106:79–93.
prolonged anti-inflammatory effect and are expected El Gazayerly ON, Hikal AH. 1997. Preparation and evaluation of
acetazolamide liposomes as an ocular delivery system. Int J Pharm
to minimize the side effects due to the selective build up
158:121–127.
of drug concentrations at the site of action. However, El Ridy MS, Khalil RM. 1999. Free versus liposome encapsulated
niosomes have proven to be more stable and were lignocaine hydrochloride topical applications. Pharmazie
of more efficacious anti-inflammatory effect compared 54:682–684.
to liposomes. Fang J, Hong C, Chiu W, Wang Y. 2001a. Effect of liposomes and
niosomes on skin permeation of enoxacin. Int J Pharm 219:61–72.
Declaration of interest: The authors report no conflicts Fang J, Yu S, Wu P, Hwang Y, Tsai Y. 2001b. In vitro skin
of interest. The authors alone are responsible for the permeation of estradiol from various proniosome formulations. Int
J Pharm 215:91–99.
content and writing of the paper. Fatouros DG, Hatzidimitriou K, Antimisiaris SG. 2001. Liposomes
encapsulating prednisolone and prednisolone-cyclodextrin
complexes: Comparison of membrane integrity and drug release.
Eur J Pharm Sci 13:287–296.
References Fresta M, Puglisi G. 1997. Corticosteroid dermal delivery with
skin-lipid liposomes. J Contr Rel 44:141–151.
Agarwal R, Katare OP, Vyas SP. 2001. Preparation and in vitro Fresta M, Ventura CA, Mezzasalma E, Puglisi G. 1998.
evaluation of liposomal/niosomal delivery systems for antipsoriatic A calorimetric study on the idebenone-phospholipid membrane
drug dithranol. Int J Pharm 228:43–52. interaction. Int J Pharm 163:133–143.
Aggarwal D, Kaur IP. 2005. Improved pharmacodynamics of timolol Friedman DI, Schwarz JS, Weisspapir M. 1995. Submicron emulsion
maleate from a mucoadhesive niosomal ophthalmic drug delivery vehicle for enhanced transdermal delivery of steroidal and
system. Int J Pharm 290:155–159. nonsteroidal anti-inflammatory drugs. J Pharm Sci 84:324–329.
Vesicularal aceclofenac systems 511

Gabriels M, Plaizier-Vercammen J. 2003. Physical and chemical Mei Z, Li X, Wu Q, Hu S, Yang X. 2005. The research on the anti-
evaluation of liposomes containing artesunate. J Pharm Biomed inflammatory activity and hepatotoxicity of triptolide- loaded solid
Anal 31:655–667. lipid nanoparticles. Pharmacol Res 51:345–351.
Ganesan MG, Weiner ND, Flynn GL, Ho NFH. 1984. Influence Moribe K, Maruyama K, Iwatsuru M. 1999. Molecular localization
of liposomal drug entrapment on percutaneous absorption. and state of Amphotericin B in PEG liposomes. Int J Pharm
Int J Pharm 20:139–154. 193:97–106.
Gianasi E, Cociancich F, Uchegbu IF, Florence AT, Duncan R. 1997. Morilla MJ, Benavidez P, Lopez MO, Bakas L, Romero EL. 2002.
Pharmaceutical and biological characterization of a doxorubicin- Development and in vitro characterization of a benznidazole
polymer conjugate (PK1) entrapped in sorbitan monstearate liposomal formulation. Int J Pharm 249:89–99.
Span 60 niosomes. Int J Pharm 148:139–148. Nagarsenker MS, Joshi AA. 1997. Preparation, characterization and
Glavas-Dodov M, Goracinova K, Mladenovska K, evaluation of liposomal dispersions of lidocaine. Drug Dev Ind
Fredro-Kumbaradzi E. 2002. Release profile of lidocaine HCl Pharm 23:1159–1165.
from topical liposomal gel formulation. Int J Pharm 242:381–384. Nagarsenker MS, Londhe VY. 2003. Preparation and evaluation of a
Guinedi AS, Mortada ND, Mansour S, Hathout RM. 2005. liposomal formulation of sodium cromoglicate. Int J Pharm
Preparation and evaluation of reverse-phase evaporation and 251:49–56.
multilamellar niosomes as ophthalmic carriers of acetazolamide. Nagarsenker MS, Londhe VY, Nadkarni GD. 1999. Preparation and
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

Int J Pharm 306:71–82. evaluation of liposomal formulations of tropicamide for ocular


Hao Y, Zhao F, Li N, Yang Y, Li K. 2002. Studies on a high delivery. Int J Pharm 190:63–71.
encapsulation of colchicine by a niosome system. Int J Pharm Namdeo A, Jain NK. 1999. Niosomal delivery of 5-fluorouracil.
244:73–80. J Microencapsulation 16:731–740.
Hathout R, Mansour S, Mortada ND, Guinedi AS. 2007. Liposomes Naresh RAR, Pillai GK, Udupa N, Chandrashekar G. 1994.
as ocular delivery system for acetzolamide: In vitro and in vivo Anti-inflammatory activity of niosome encapsulated diclofenac
studies. AAPS Pharm Sci Tech 8:article E1–E2. sodium in arthritic rats. Indian J Pharmacol 26:46–48.
Insel PA. 1992. Analgesic-antipyretics and anti-inflammatory agents: Nasseri B. 2005. Effect of cholesterol and temperature on the elastic
Drugs employed in the treatment of rheumatoid arthritis and gout. properties of niosomal membranes. Int J Pharm 300:95–101.
In: Goodman LS, Gilman A, Rall TW, Nies AS, Taylor P, editors. Ozer AY, Talsama H. 1989. Preparation and stability of liposomes
The pharmacological basis of therapeutics. New York: containing 5-fluorouracil. Int J Pharm 55:185–191.
McGraw-Hill International editions. pp 638–681. Padamwar MN, Pokharkar VB. 2006. Development of vitamin
Joshi MR, Misra A. 2001. Liposomal budesonide for dry powder loaded topical liposomal formulation using factorial
inhaler: preparation and stabilization. AAPS Pharm Sci Tech design approach: Drug deposition and stability. Int J Pharm
For personal use only.

2:article 25 44–53. 320:37–44.


Junginger HE, Hofland HEJ, Bouwstra JA. 1991. Liposomes and Pasero G, Marcolongo R, Serni U, Parnham MJ, Ferrer F. 1995.
niosomes interactions with human skin. Cosmetics Toiletries Multi-center, double blind comparative study of the efficacy and
106:45–50. safety of aceclofenac and diclofenac in the treatment of rheumatoid
Kulkarni SB, Betageri GV, Singh M. 1995. Factors affecting arthritis. Curr Med Res Opin 13:305–315.
microencapsulation of drugs in liposomes. J Microencapsulation Patel VB, Misra AN. 1999. Encapsulation and stability of clofazimine
12:229–246. liposomes. J Microencapsulation 16:357–367.
Law SL, Lo WY, Lin M. 1994. Increase of liposome stability by Pavelic Z, Skalko-Basnet N, Schubert R. 2001. Liposomal gels for
incorporation of bovine serum albumin. Drug Dev Ind Pharm vaginal drug delivery. Int J Pharm 219:139–149.
20:1411–1423. Penzes T, Blazso G, Aigner Z, Falkay G, Eros I. 2005. Topical
Maestrelli F, Gonzalez-Rodrjguez ML, Rabasco AM, Mura P. 2005. absorption of piroxicam from organogels—in vitro and in vivo
Preparation and characterization of liposomes encapsulating correlations. Int J Pharm 298:47–54.
ketoprofen-cyclodextrin complexes for transdermal drug delivery. Pereira-Lachataignerais J, Pons R, Panizza P, Courbin L, Rouch J,
Int J Pharm 298:55–67. Lopez O. 2006. Study and formation of vesicle systems with low
Maghraby GMME, Campbell M, Finnin BC. 2005. Mechansims of polydispersity index by ultrasound method. Chem Phys Lipids
action of novel skin lipid liposomes. Int J Pharm 305:90–104. 140:88–97.
Manconi M, Sinico C, Valenti D, Loy G, Fadda AM. 2002. Niosomes Rogerson A, Cummings J, Florence AT. 1987. Adriamycin-loaded
as carriers for tretinoin I. Preapartion and properties. Int J Pharm niosomes-drug entrapment, stability and release.
234:237–248. J Microencapsulation 4:321–328.
Manconi M, Valenti D, Sinico C, Lai F, Loy G, Fadda AM. 2003. Rolland A. 1993. Particulate carriers in dermal and transdermal drug
Niosomes as carriers for tretinoin II. Influence of vesicular delivery: Myth or reality?. In: Rolland A, editor. Pharmaceutical
incorporation on tretinoin photostability. Int J Pharm particulate carriers: Therapeutic applications. New York: Marcel
260:261–272. Dekker. pp 367–421.
Manosroi A, Kongkaneramit L, Manosroi J. 2004. Stability and Ruckmani K, Jayakar B, Ghosal SK. 2000. Nonionic surfactant
transdermal absorption of topical Amphotericin B liposome vesicles (Niosomes) of cytarabine hydrochloride for effective
formulations. Int J Pharm 270:279–286. treatment of leukemias: Encapsulation, storage and in vitro release.
Manosroi A, Podjanasoonthon K, Manosroi J. 2002. Development of Drug Dev Ind Pharm 26:217–222.
novel topical tranexamic acid liposome formulations. Int J Pharm Shahiwala A, Misra A. 2002. Studies in topical application of
235:61–70. niosomally entrapped nimesulide. J Pharm Pharm Sci 5:220–225.
Manosroi A, Wongtrakul P, Manosroi J, Sakai H, Sugawara F, Sharma BB, Jain SK, Vyas SP. 1994. Topical liposome system bearing
Yuasa M, Abe M. 2003. Characterization of vesicles prepared with local anaesthetic lignocaine: Preparation and evaluation.
various non ionic surfactants mixed with cholesterol. Coll Surf B J Microencapsulation 11:279–286.
Biointerfaces 30:129–138. Sihorkar V, Vyas SP. 2000. Polysaccharide coated niosomes for oral
Martel-Pelletier J, Cloutier JM, Pelletier JP. 1997. Effects of drug delivery formulation and in vitro stability studies. Pharmazie
aceclofenac and diclofenac on synovial inflammatory factors in 55:107–113.
human osteoarthritis. Clin Drug Invest 14:226–232. Sinico C, De Logu A, Lai F, Valenti D, Manconi M, Loy G,
Mei Z, Chen H, Weng T, Yang Y, Yang X. 2003. Solid lipid Bonsignore L, Fadda AM. 2005. Liposomal incorporation of
nanoparticles and microemulsion for topical delivery of triptolide. Artemisia arboscens L. essential oil and in vitro antiviral activity.
Eur J Pharm Biopharm 56:189–196. Eur J Pharm Biopharm 59:161–168.
512 M. Nasr et al.

Skalko-Basnet N, Pavelic Z, Becirevic-Lacan M. 2000. Liposomes Vyas SP, Kannan ME, Jain S, Mishra V, Singh P. 2004. Design of
containing drug and cyclodextrin prepared by the one-step spray- liposomal aerosols for improved delivery of rifampicin to alveolar
drying method. Drug Dev Ind Pharm 26:1279–1284. macrophages. Int J Pharm 269:37–49.
Tabbakhian M, Tavakoli N, Jaafari MR, Daneshamouz S. 2006. Vyas SP, Singh R, Asati RK. 1995. Liposomally encapsulated
Enhancement of follicular delivery of finasteride by liposomes and diclofenac for sonophoresis induced systemic delivery.
niosomes 1. In vitro permeation and in vivo deposition studies using J Microencapsulation 12:149–154.
hamster flank and ear models. Int J Pharm 323:1–10. Vyas SP, Venkatesan N. 1999. Poly (pthaloyl-L-lysine)-coated multi-
Taylor KMG, Taylor G, Kellaway IW, Stevens J. 1990. Drug lamellar vesicles for controlled drug delivery: In vitro and in vivo
entrapment and release from multilamellar and reverse-phase performance evaluation. Pharm Acta Helv 74:51–58.
evaporation liposomes. Int J Pharm 58:49–55. Yan-yu X, Yun-mei S, Zhi-peng C, Qi-neng P. 2006. Prepartion of
Uchegbu IF, Duncan R. 1997. Niosomes containing N-(2-hydro- silymarin proliposome: A new way to increase oral bioavailability
xypropyl) methacrylamide copolymer- doxorubicin (PK1): Effect of silymarin in beagle dogs. Int J Pharm 319:162–168.
of method of preparation and choice of surfactant on niosome Yoshioka T, Sternberg B, Florence AT. 1994. Preparation
characteristics and a preliminary study of body distribution. and properties of vesicles (niosomes) of sorbitan monoesters
Int J Pharm 155:7–17. (Span 20, 40, 60 and 80) and sorbitan triester (Span 85). Int J
Uchegbu IF, Florence AT. 1995. Non-ionic surfactant vesicles Pharm 105:1–6.
Journal of Microencapsulation Downloaded from informahealthcare.com by University of Calgary on 03/18/13

(niosomes): Physical and pharmaceutical chemistry. Adv Coll Zeisig R, Shimada K, Hirota S, Arndt D. 1996. Effect of
Interf Sci 58:1–55. sterical stabilization on macrophage uptake in vitro and on
Uchegbu IF, Vyas SP. 1998. Non Ionic surfactant based vesicles thickness of the fixed aqueous layer of liposomes made from
(niosomes) in drug delivery. Int J Pharm 172:33–70. alkylphosphocholines. Biochim Biophys Acta Biomembranes
Vora B, Khopade AJ, Jain NK. 1998. Proniosome based transdermal 1285:237–245.
delivery of levonorgesterol for effective contraception. J Contr Rel Zhang JH, Zhu JB. 1999. A novel method to prepare liposomes
54:149–165. containing amikacin. J Microencapsulation 16:511–516.
For personal use only.

S-ar putea să vă placă și